1
|
Morales EA, Fitz GN, Tyska MJ. Mitotic spindle positioning protein (MISP) preferentially binds to aged F-actin. J Biol Chem 2024; 300:107279. [PMID: 38588808 PMCID: PMC11101845 DOI: 10.1016/j.jbc.2024.107279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 03/14/2024] [Accepted: 04/01/2024] [Indexed: 04/10/2024] Open
Abstract
Actin bundling proteins crosslink filaments into polarized structures that shape and support membrane protrusions including filopodia, microvilli, and stereocilia. In the case of epithelial microvilli, mitotic spindle positioning protein (MISP) is an actin bundler that localizes specifically to the basal rootlets, where the pointed ends of core bundle filaments converge. Previous studies established that MISP is prevented from binding more distal segments of the core bundle by competition with other actin-binding proteins. Yet whether MISP holds a preference for binding directly to rootlet actin remains an open question. By immunostaining native intestinal tissue sections, we found that microvillar rootlets are decorated with the severing protein, cofilin, suggesting high levels of ADP-actin in these structures. Using total internal reflection fluorescence microscopy assays, we also found that purified MISP exhibits a binding preference for ADP- versus ADP-Pi-actin-containing filaments. Consistent with this, assays with actively growing actin filaments revealed that MISP binds at or near their pointed ends. Moreover, although substrate attached MISP assembles filament bundles in parallel and antiparallel configurations, in solution MISP assembles parallel bundles consisting of multiple filaments exhibiting uniform polarity. These discoveries highlight nucleotide state sensing as a mechanism for sorting actin bundlers along filaments and driving their accumulation near filament ends. Such localized binding might drive parallel bundle formation and/or locally modulate bundle mechanical properties in microvilli and related protrusions.
Collapse
Affiliation(s)
- E Angelo Morales
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Gillian N Fitz
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA.
| |
Collapse
|
2
|
Heyn JCJ, Rädler JO, Falcke M. Mesenchymal cell migration on one-dimensional micropatterns. Front Cell Dev Biol 2024; 12:1352279. [PMID: 38694822 PMCID: PMC11062138 DOI: 10.3389/fcell.2024.1352279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/29/2024] [Indexed: 05/04/2024] Open
Abstract
Quantitative studies of mesenchymal cell motion are important to elucidate cytoskeleton function and mechanisms of cell migration. To this end, confinement of cell motion to one dimension (1D) significantly simplifies the problem of cell shape in experimental and theoretical investigations. Here we review 1D migration assays employing micro-fabricated lanes and reflect on the advantages of such platforms. Data are analyzed using biophysical models of cell migration that reproduce the rich scenario of morphodynamic behavior found in 1D. We describe basic model assumptions and model behavior. It appears that mechanical models explain the occurrence of universal relations conserved across different cell lines such as the adhesion-velocity relation and the universal correlation between speed and persistence (UCSP). We highlight the unique opportunity of reproducible and standardized 1D assays to validate theory based on statistical measures from large data of trajectories and discuss the potential of experimental settings embedding controlled perturbations to probe response in migratory behavior.
Collapse
Affiliation(s)
- Johannes C. J. Heyn
- Fakultät für Physik, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Joachim O. Rädler
- Fakultät für Physik, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Martin Falcke
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Physics, Humboldt University, Berlin, Germany
| |
Collapse
|
3
|
Goode BL, Eskin J, Shekhar S. Mechanisms of actin disassembly and turnover. J Cell Biol 2023; 222:e202309021. [PMID: 37948068 PMCID: PMC10638096 DOI: 10.1083/jcb.202309021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Cellular actin networks exhibit a wide range of sizes, shapes, and architectures tailored to their biological roles. Once assembled, these filamentous networks are either maintained in a state of polarized turnover or induced to undergo net disassembly. Further, the rates at which the networks are turned over and/or dismantled can vary greatly, from seconds to minutes to hours or even days. Here, we review the molecular machinery and mechanisms employed in cells to drive the disassembly and turnover of actin networks. In particular, we highlight recent discoveries showing that specific combinations of conserved actin disassembly-promoting proteins (cofilin, GMF, twinfilin, Srv2/CAP, coronin, AIP1, capping protein, and profilin) work in concert to debranch, sever, cap, and depolymerize actin filaments, and to recharge actin monomers for new rounds of assembly.
Collapse
Affiliation(s)
- Bruce L. Goode
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA, USA
| | - Julian Eskin
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA, USA
| | - Shashank Shekhar
- Departments of Physics, Cell Biology and Biochemistry, Emory University, Atlanta, GA, USA
| |
Collapse
|
4
|
Towsif EM, Shekhar S. Cyclase-associated protein is a pro-formin anti-capping processive depolymerase of actin barbed and pointed ends. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.30.569482. [PMID: 38076850 PMCID: PMC10705416 DOI: 10.1101/2023.11.30.569482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Cellular actin networks display distinct assembly and disassembly dynamics resulting from multicomponent reactions occurring primarily at the two ends and the sides of actin filaments [1-3]. While barbed ends are considered the hotspot of actin assembly [4], disassembly is thought to primarily occur via reactions on filament sides and pointed ends [3, 5-11]. Cyclase-associated protein (CAP) has emerged as the main protagonist of actin disassembly and remodeling - it collaborates with cofilin to increase pointed-end depolymerization by 300-fold [6, 7], promotes filament "coalescence" in presence of Abp1 [12], and accelerates nucleotide exchange to regenerate monomers for new rounds of assembly [13-15]. CAP has also been reported to enhance cofilin-mediated severing [16, 17], but these claims have since been challenged [7]. Using microfluidics-assisted three-color single-molecule imaging, we now reveal that CAP also has important functions at filament barbed ends. We reveal that CAP is a processive barbed-end depolymerase capable of tracking both ends of the filament. Each CAP binding event leads to removal of about 5,175 and 620 subunits from the barbed and pointed ends respectively. We find that the WH2 domain is essential, and the CARP domain is dispensable for barbed-end depolymerization. We show that CAP co-localizes with barbed-end bound formin and capping protein, in the process increasing residence time of formin by 10-fold and promoting dissociation of CP by 4-fold. Our barbed-end observations combined with previously reported activities of CAP at pointed ends and sides, firmly establish CAP as a key player in actin dynamics.
Collapse
Affiliation(s)
- Ekram M. Towsif
- Departments of Physics, Cell biology and Biochemistry, Emory University, Atlanta, GA 30322, USA
| | - Shashank Shekhar
- Departments of Physics, Cell biology and Biochemistry, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
5
|
Morales EA, Tyska MJ. Mitotic spindle positioning protein (MISP) is an actin bundler that senses ADP-actin and binds near the pointed ends of filaments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.05.539649. [PMID: 37205433 PMCID: PMC10187293 DOI: 10.1101/2023.05.05.539649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Actin bundling proteins crosslink filaments into polarized structures that shape and support membrane protrusions including filopodia, microvilli, and stereocilia. In the case of epithelial microvilli, mitotic spindle positioning protein (MISP) is an actin bundler that localizes specifically to the basal rootlets, where the pointed ends of core bundle filaments converge. Previous studies established that MISP is prevented from binding more distal segments of the core bundle by competition with other actin binding proteins. Yet whether MISP holds a preference for binding directly to rootlet actin remains an open question. Using in vitro TIRF microscopy assays, we found that MISP exhibits a clear binding preference for filaments enriched in ADP-actin monomers. Consistent with this, assays with actively growing actin filaments revealed that MISP binds at or near their pointed ends. Moreover, although substrate attached MISP assembles filament bundles in parallel and antiparallel configurations, in solution MISP assembles parallel bundles consisting of multiple filaments exhibiting uniform polarity. These discoveries highlight nucleotide state sensing as a mechanism for sorting actin bundlers along filaments and driving their accumulation near filament ends. Such localized binding might drive parallel bundle formation and/or locally modulate bundle mechanical properties in microvilli and related protrusions.
Collapse
|
6
|
Al-Hiyasat A, Tuna Y, Kuo YW, Howard J. Herding of proteins by the ends of shrinking polymers. Phys Rev E 2023; 107:L042601. [PMID: 37198784 PMCID: PMC11550390 DOI: 10.1103/physreve.107.l042601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/17/2023] [Indexed: 05/19/2023]
Abstract
The control of biopolymer length is mediated by proteins that localize to polymer ends and regulate polymerization dynamics. Several mechanisms have been proposed to achieve end localization. Here, we propose a novel mechanism by which a protein that binds to a shrinking polymer and slows its shrinkage will be spontaneously enriched at the shrinking end through a "herding" effect. We formalize this process using both lattice-gas and continuum descriptions, and we present experimental evidence that the microtubule regulator spastin employs this mechanism. Our findings extend to more general problems involving diffusion within shrinking domains.
Collapse
Affiliation(s)
- Amer Al-Hiyasat
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06511, USA
- Department of Physics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Yazgan Tuna
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06511, USA
| | - Yin-Wei Kuo
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06511, USA
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06511, USA
| |
Collapse
|
7
|
Colin A, Kotila T, Guérin C, Orhant-Prioux M, Vianay B, Mogilner A, Lappalainen P, Théry M, Blanchoin L. Recycling of the actin monomer pool limits the lifetime of network turnover. EMBO J 2023; 42:e112717. [PMID: 36912152 PMCID: PMC10152149 DOI: 10.15252/embj.2022112717] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 03/14/2023] Open
Abstract
Intracellular organization is largely mediated by actin turnover. Cellular actin networks continuously assemble and disassemble, while maintaining their overall appearance. This behavior, called "dynamic steady state," allows cells to sense and adapt to their environment. However, how structural stability can be maintained during the constant turnover of a limited actin monomer pool is poorly understood. To answer this question, we developed an experimental system where polystyrene beads are propelled by an actin comet in a microwell containing a limited amount of components. We used the speed and the size of the actin comet tails to evaluate the system's monomer consumption and its lifetime. We established the relative contribution of actin assembly, disassembly, and recycling for a bead movement over tens of hours. Recycling mediated by cyclase-associated protein (CAP) is the key step in allowing the reuse of monomers for multiple assembly cycles. ATP supply and protein aging are also factors that limit the lifetime of actin turnover. This work reveals the balancing mechanism for long-term network assembly with a limited amount of building blocks.
Collapse
Affiliation(s)
- Alexandra Colin
- CytoMorpho Lab, Laboratoire de Physiologie Cellulaire & Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, Grenoble, France
| | - Tommi Kotila
- Institute of Biotechnology and Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Christophe Guérin
- CytoMorpho Lab, Laboratoire de Physiologie Cellulaire & Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, Grenoble, France
| | - Magali Orhant-Prioux
- CytoMorpho Lab, Laboratoire de Physiologie Cellulaire & Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, Grenoble, France
| | - Benoit Vianay
- CytoMorpho Lab, Institut de Recherche Saint Louis, U976 Human Immunology Pathophysiology Immunotherapy (HIPI), University of Paris, INSERM, CEA, Paris, France
| | - Alex Mogilner
- Courant Institute of Mathematical Sciences, New York University, New York, NY, USA.,Department of Biology, New York University, New York, NY, USA
| | - Pekka Lappalainen
- Institute of Biotechnology and Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Manuel Théry
- CytoMorpho Lab, Laboratoire de Physiologie Cellulaire & Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, Grenoble, France.,CytoMorpho Lab, Institut de Recherche Saint Louis, U976 Human Immunology Pathophysiology Immunotherapy (HIPI), University of Paris, INSERM, CEA, Paris, France
| | - Laurent Blanchoin
- CytoMorpho Lab, Laboratoire de Physiologie Cellulaire & Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, Grenoble, France.,CytoMorpho Lab, Institut de Recherche Saint Louis, U976 Human Immunology Pathophysiology Immunotherapy (HIPI), University of Paris, INSERM, CEA, Paris, France
| |
Collapse
|
8
|
Banerjee DS, Banerjee S. Emergence and maintenance of variable-length actin filaments in a limiting pool of building blocks. Biophys J 2022; 121:2436-2448. [PMID: 35598045 DOI: 10.1016/j.bpj.2022.05.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 04/10/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Actin is one of the key structural components of the eukaryotic cytoskeleton that regulates cellular architecture and mechanical properties. Dynamic regulation of actin filament length and organization is essential for the control of many physiological processes including cell adhesion, motility and division. While previous studies have mostly focused on the mechanisms controlling the mean length of individual actin filaments, it remains poorly understood how distinct actin filament populations in cells maintain different lengths using the same set of molecular building blocks. Here we develop a theoretical model for the length regulation of multiple actin filaments by nucleation and growth rate modulation by actin binding proteins in a limiting pool of monomers. We first show that spontaneous nucleation of actin filaments naturally leads to heterogeneities in filament length distribution. We then investigate the effects of filament growth inhibition by capping proteins and growth promotion by formin proteins on filament length distribution. We find that filament length heterogeneity can be increased by growth inhibition, whereas growth promoters do not significantly affect length heterogeneity. Interestingly, a competition between filament growth inhibitors and growth promoters can give rise to bimodal filament length distribution as well as a highly heterogeneous length distribution with large statistical dispersion. We quantitatively predict how heterogeneity in actin filament length can be modulated by tuning F-actin nucleation and growth rates in order to create distinct filament subpopulations with different lengths. SIGNIFICANCE: Actin filaments organize into different functional network architectures within eukaryotic cells. To maintain distinct actin network architectures, it is essential to regulate the lengths of actin filaments. While the mechanisms controlling the lengths of individual actin filaments have been extensively studied, the regulation of length heterogeneity in actin filament populations is not well understood. Here we show that the modulation of actin filament growth and nucleation rates by actin binding proteins can regulate actin length distribution and create distinct sub-populations with different lengths. In particular, by tuning concentrations of formin, profilin and capping proteins, various aspects of actin filament length distribution can be controlled. Insights gained from our results may have significant implications for the regulation of actin filament length heterogeneity and architecture within a cell.
Collapse
Affiliation(s)
- Deb Sankar Banerjee
- Carnegie Mellon University, Department of Physics, Pittsburgh, PA 15213, USA
| | - Shiladitya Banerjee
- Carnegie Mellon University, Department of Physics, Pittsburgh, PA 15213, USA
| |
Collapse
|
9
|
Jiang CF, Sun YM. Label-free monitoring of spatiotemporal changes in the stem cell cytoskeletons in time-lapse phase-contrast microscopy. BIOMEDICAL OPTICS EXPRESS 2022; 13:2323-2333. [PMID: 35519244 PMCID: PMC9045902 DOI: 10.1364/boe.452822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 06/14/2023]
Abstract
Investigation of the dynamic structural changes in the actin cytoskeleton during cell migration provides crucial information about the physiological conditions of a stem cell during in-vitro culture. Here we proposed a quantitative analytical model associated with texture extraction with cell tracking techniques for in situ monitoring of the cytoskeletal density change of stem cells in phase-contrast microscopy without fluorescence staining. The reliability of the model in quantifying the texture density with different orientation was first validated using a series of simulated textural images. The capability of the method to reflect the spatiotemporal regulation of the cytoskeletal structure of a living stem cell was further proved by applying it to a set of 72 h phase-contrast microscopic video of the growth dynamics of mesenchymal stem cells in vitro culture.
Collapse
Affiliation(s)
- Ching-Fen Jiang
- Graduate Degree Program of Smart Healthcare & Bioinformatics, I-Shou University, Kaohsiung, Taiwan
| | - Yu-Man Sun
- Department of Biomedical Engineering, I-Shou University, Kaohsiung, Taiwan
| |
Collapse
|
10
|
Chinowsky CR, Pinette JA, Meenderink LM, Lau KS, Tyska MJ. Nonmuscle myosin-2 contractility-dependent actin turnover limits the length of epithelial microvilli. Mol Biol Cell 2020; 31:2803-2815. [PMID: 33026933 PMCID: PMC7851865 DOI: 10.1091/mbc.e20-09-0582] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022] Open
Abstract
Brush border microvilli enable functions that are critical for epithelial homeostasis, including solute uptake and host defense. However, the mechanisms that regulate the assembly and morphology of these protrusions are poorly understood. The parallel actin bundles that support microvilli have their pointed-end rootlets anchored in a filamentous meshwork referred to as the "terminal web." Although classic electron microscopy studies revealed complex ultrastructure, the composition and function of the terminal web remain unclear. Here we identify nonmuscle myosin-2C (NM2C) as a component of the terminal web. NM2C is found in a dense, isotropic layer of puncta across the subapical domain, which transects the rootlets of microvillar actin bundles. Puncta are separated by ∼210 nm, the expected size of filaments formed by NM2C. In intestinal organoid cultures, the terminal web NM2C network is highly dynamic and exhibits continuous remodeling. Using pharmacological and genetic perturbations in cultured intestinal epithelial cells, we found that NM2C controls the length of growing microvilli by regulating actin turnover in a manner that requires a fully active motor domain. Our findings answer a decades-old question on the function of terminal web myosin and hold broad implications for understanding apical morphogenesis in diverse epithelial systems.
Collapse
Affiliation(s)
- Colbie R Chinowsky
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Julia A Pinette
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Leslie M Meenderink
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Ken S Lau
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
11
|
Allen GM, Lee KC, Barnhart EL, Tsuchida MA, Wilson CA, Gutierrez E, Groisman A, Theriot JA, Mogilner A. Cell Mechanics at the Rear Act to Steer the Direction of Cell Migration. Cell Syst 2020; 11:286-299.e4. [PMID: 32916096 PMCID: PMC7530145 DOI: 10.1016/j.cels.2020.08.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/17/2020] [Accepted: 08/10/2020] [Indexed: 01/18/2023]
Abstract
Motile cells navigate complex environments by changing their direction of travel, generating left-right asymmetries in their mechanical subsystems to physically turn. Currently, little is known about how external directional cues are propagated along the length scale of the whole cell and integrated with its force-generating apparatus to steer migration mechanically. We examine the mechanics of spontaneous cell turning in fish epidermal keratocytes and find that the mechanical asymmetries responsible for turning behavior predominate at the rear of the cell, where there is asymmetric centripetal actin flow. Using experimental perturbations, we identify two linked feedback loops connecting myosin II contractility, adhesion strength and actin network flow in turning cells that are sufficient to explain the observed cell shapes and trajectories. Notably, asymmetries in actin polymerization at the cell leading edge play only a minor role in the mechanics of cell turning-that is, cells steer from the rear.
Collapse
Affiliation(s)
- Greg M Allen
- Department of Biochemistry and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kun Chun Lee
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, USA
| | - Erin L Barnhart
- Department of Biochemistry and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark A Tsuchida
- Department of Biochemistry and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Cyrus A Wilson
- Department of Biochemistry and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Edgar Gutierrez
- Department of Physics, University of California, San Diego, San Diego, CA 92023, USA
| | - Alexander Groisman
- Department of Physics, University of California, San Diego, San Diego, CA 92023, USA
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.
| | - Alex Mogilner
- Courant Institute of Mathematical Sciences and Department of Biology, New York University, New York, NY 10012, USA.
| |
Collapse
|
12
|
Caruso S, Atkin-Smith GK, Baxter AA, Tixeira R, Jiang L, Ozkocak DC, Santavanond JP, Hulett MD, Lock P, Phan TK, Poon IKH. Defining the role of cytoskeletal components in the formation of apoptopodia and apoptotic bodies during apoptosis. Apoptosis 2020; 24:862-877. [PMID: 31489517 DOI: 10.1007/s10495-019-01565-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
During apoptosis, dying cells undergo dynamic morphological changes that ultimately lead to their disassembly into fragments called apoptotic bodies (ApoBDs). Reorganisation of the cytoskeletal structures is key in driving various apoptotic morphologies, including the loss of cell adhesion and membrane bleb formation. However, whether cytoskeletal components are also involved in morphological changes that occur later during apoptosis, such as the recently described generation of thin apoptotic membrane protrusions called apoptopodia and subsequent ApoBD formation, is not well defined. Through monitoring the progression of apoptosis by confocal microscopy, specifically focusing on the apoptopodia formation step, we characterised the presence of F-actin and microtubules in a subset of apoptopodia generated by T cells and monocytes. Interestingly, targeting actin polymerisation and microtubule assembly pharmacologically had no major effect on apoptopodia formation. These data demonstrate apoptopodia as a novel type of membrane protrusion that could be formed in the absence of actin polymerisation and microtubule assembly.
Collapse
Affiliation(s)
- Sarah Caruso
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Georgia K Atkin-Smith
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Amy A Baxter
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Rochelle Tixeira
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Lanzhou Jiang
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Dilara C Ozkocak
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Jascinta P Santavanond
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Mark D Hulett
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Peter Lock
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Thanh Kha Phan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Ivan K H Poon
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
13
|
Alexandrova AY, Chikina AS, Svitkina TM. Actin cytoskeleton in mesenchymal-to-amoeboid transition of cancer cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:197-256. [PMID: 33066874 DOI: 10.1016/bs.ircmb.2020.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During development of metastasis, tumor cells migrate through different tissues and encounter different extracellular matrices. An ability of cells to adapt mechanisms of their migration to these diverse environmental conditions, called migration plasticity, gives tumor cells an advantage over normal cells for long distant dissemination. Different modes of individual cell motility-mesenchymal and amoeboid-are driven by different molecular mechanisms, which largely depend on functions of the actin cytoskeleton that can be modulated in a wide range by cellular signaling mechanisms in response to environmental conditions. Various triggers can switch one motility mode to another, but regulations of these transitions are incompletely understood. However, understanding of the mechanisms driving migration plasticity is instrumental for finding anti-cancer treatment capable to stop cancer metastasis. In this review, we discuss cytoskeletal features, which allow the individually migrating cells to switch between mesenchymal and amoeboid migrating modes, called mesenchymal-to-amoeboid transition (MAT). We briefly describe main characteristics of different cell migration modes, and then discuss the triggering factors that initiate MAT with special attention to cytoskeletal features essential for migration plasticity.
Collapse
Affiliation(s)
- Antonina Y Alexandrova
- Laboratory of Mechanisms of Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia.
| | - Aleksandra S Chikina
- Cell Migration and Invasion and Spatio-Temporal Regulation of Antigen Presentation teams, UMR144/U932 Institut Curie, Paris, France
| | - Tatyana M Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
14
|
Jaumouillé V, Waterman CM. Physical Constraints and Forces Involved in Phagocytosis. Front Immunol 2020; 11:1097. [PMID: 32595635 PMCID: PMC7304309 DOI: 10.3389/fimmu.2020.01097] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/06/2020] [Indexed: 01/02/2023] Open
Abstract
Phagocytosis is a specialized process that enables cellular ingestion and clearance of microbes, dead cells and tissue debris that are too large for other endocytic routes. As such, it is an essential component of tissue homeostasis and the innate immune response, and also provides a link to the adaptive immune response. However, ingestion of large particulate materials represents a monumental task for phagocytic cells. It requires profound reorganization of the cell morphology around the target in a controlled manner, which is limited by biophysical constraints. Experimental and theoretical studies have identified critical aspects associated with the interconnected biophysical properties of the receptors, the membrane, and the actin cytoskeleton that can determine the success of large particle internalization. In this review, we will discuss the major physical constraints involved in the formation of a phagosome. Focusing on two of the most-studied types of phagocytic receptors, the Fcγ receptors and the complement receptor 3 (αMβ2 integrin), we will describe the complex molecular mechanisms employed by phagocytes to overcome these physical constraints.
Collapse
Affiliation(s)
- Valentin Jaumouillé
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Clare M Waterman
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
15
|
Mogilner A, Barnhart EL, Keren K. Experiment, theory, and the keratocyte: An ode to a simple model for cell motility. Semin Cell Dev Biol 2020; 100:143-151. [DOI: 10.1016/j.semcdb.2019.10.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/27/2019] [Accepted: 10/31/2019] [Indexed: 01/20/2023]
|
16
|
Cantini M, Donnelly H, Dalby MJ, Salmeron‐Sanchez M. The Plot Thickens: The Emerging Role of Matrix Viscosity in Cell Mechanotransduction. Adv Healthc Mater 2020; 9:e1901259. [PMID: 31815372 DOI: 10.1002/adhm.201901259] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/26/2019] [Indexed: 12/21/2022]
Abstract
Cell mechanotransduction is an area of intense research focus. Until now, very limited tools have existed to study how cells respond to changes in the extracellular matrix beyond, for example, mechanical deformation studies and twisting cytometry. However, emerging are a range of elastic, viscoelastic and even purely viscous materials that deform and dissipate on cellular length and timescales. This article reviews developments in these materials, typically translating from 2D model surfaces to 3D microenvironments and explores how cells interact with them. Specifically, it focuses on emerging concepts such as the molecular clutch model, how different extracellular matrix proteins engage the clutch under viscoelastic-stress relaxation conditions, and how mechanotransduction can drive transcriptional control through regulators such as YAP/TAZ.
Collapse
Affiliation(s)
- Marco Cantini
- Centre for the Cellular MicroenvironmentUniversity of Glasgow Glasgow G12 8QQ UK
| | - Hannah Donnelly
- Centre for the Cellular MicroenvironmentUniversity of Glasgow Glasgow G12 8QQ UK
| | - Matthew J. Dalby
- Centre for the Cellular MicroenvironmentUniversity of Glasgow Glasgow G12 8QQ UK
| | | |
Collapse
|
17
|
Abstract
Integrins, and integrin-mediated adhesions, have long been recognized to provide the main molecular link attaching cells to the extracellular matrix (ECM) and to serve as bidirectional hubs transmitting signals between cells and their environment. Recent evidence has shown that their combined biochemical and mechanical properties also allow integrins to sense, respond to and interact with ECM of differing properties with exquisite specificity. Here, we review this work first by providing an overview of how integrin function is regulated from both a biochemical and a mechanical perspective, affecting integrin cell-surface availability, binding properties, activation or clustering. Then, we address how this biomechanical regulation allows integrins to respond to different ECM physicochemical properties and signals, such as rigidity, composition and spatial distribution. Finally, we discuss the importance of this sensing for major cell functions by taking cell migration and cancer as examples.
Collapse
|
18
|
Garner RM, Skariah G, Hadjitheodorou A, Belliveau NM, Savinov A, Footer MJ, Theriot JA. Neutrophil-like HL-60 cells expressing only GFP-tagged β-actin exhibit nearly normal motility. Cytoskeleton (Hoboken) 2020; 77:181-196. [PMID: 32072765 PMCID: PMC7383899 DOI: 10.1002/cm.21603] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/21/2019] [Accepted: 01/27/2020] [Indexed: 12/30/2022]
Abstract
Observations of actin dynamics in living cells using fluorescence microscopy have been foundational in the exploration of the mechanisms underlying cell migration. We used CRISPR/Cas9 gene editing to generate neutrophil‐like HL‐60 cell lines expressing GFP‐β‐actin from the endogenous locus (ACTB). In light of many previous reports outlining functional deficiencies of labeled actin, we anticipated that HL‐60 cells would only tolerate a monoallelic edit, as biallelic edited cells would produce no normal β‐actin. Surprisingly, we recovered viable monoallelic GFP‐β‐actin cells as well as biallelic edited GFP‐β‐actin cells, in which one copy of the ACTB gene is silenced and the other contains the GFP tag. Furthermore, the edited cells migrate with similar speeds and persistence as unmodified cells in a variety of motility assays, and have nearly normal cell shapes. These results might partially be explained by our observation that GFP‐β‐actin incorporates into the F‐actin network in biallelic edited cells at similar efficiencies as normal β‐actin in unedited cells. Additionally, the edited cells significantly upregulate γ‐actin, perhaps helping to compensate for the loss of normal β‐actin. Interestingly, biallelic edited cells have only modest changes in global gene expression relative to the monoallelic line, as measured by RNA sequencing. While monoallelic edited cells downregulate expression of the tagged allele and are thus only weakly fluorescent, biallelic edited cells are quite bright and well‐suited for live cell microscopy. The nondisruptive phenotype and direct interpretability of this fluorescent tagging approach make it a promising tool for studying actin dynamics in these rapidly migrating and highly phagocytic cells.
Collapse
Affiliation(s)
- Rikki M Garner
- Biophysics Program, Stanford University School of Medicine, Stanford, CA.,Department of Biology, Howard Hughes Medical Institute, University of Washington, Seattle, WA
| | - Gemini Skariah
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA
| | - Amalia Hadjitheodorou
- Department of Bioengineering, Stanford University Schools of Medicine and Engineering, Stanford, CA
| | - Nathan M Belliveau
- Department of Biology, Howard Hughes Medical Institute, University of Washington, Seattle, WA
| | - Andrew Savinov
- Department of Genome Sciences, University of Washington, Seattle, WA
| | - Matthew J Footer
- Department of Biology, Howard Hughes Medical Institute, University of Washington, Seattle, WA
| | - Julie A Theriot
- Department of Biology, Howard Hughes Medical Institute, University of Washington, Seattle, WA
| |
Collapse
|
19
|
Abstract
The polymerization-depolymerization dynamics of cytoskeletal proteins play essential roles in the self-organization of cytoskeletal structures, in eukaryotic as well as prokaryotic cells. While advances in fluorescence microscopy and in vitro reconstitution experiments have helped to study the dynamic properties of these complex systems, methods that allow to collect and analyze large quantitative datasets of the underlying polymer dynamics are still missing. Here, we present a novel image analysis workflow to study polymerization dynamics of active filaments in a nonbiased, highly automated manner. Using treadmilling filaments of the bacterial tubulin FtsZ as an example, we demonstrate that our method is able to specifically detect, track and analyze growth and shrinkage of polymers, even in dense networks of filaments. We believe that this automated method can facilitate the analysis of a large variety of dynamic cytoskeletal systems, using standard time-lapse movies obtained from experiments in vitro as well as in the living cell. Moreover, we provide scripts implementing this method as supplementary material.
Collapse
|
20
|
Tilve S, Iweka CA, Bao J, Hawken N, Mencio CP, Geller HM. Phospholipid phosphatase related 1 (PLPPR1) increases cell adhesion through modulation of Rac1 activity. Exp Cell Res 2020; 389:111911. [PMID: 32061832 DOI: 10.1016/j.yexcr.2020.111911] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/03/2020] [Accepted: 02/11/2020] [Indexed: 12/31/2022]
Abstract
Phospholipid Phosphatase-Related Protein Type 1 (PLPPR1) is a six-transmembrane protein that belongs to the family of plasticity-related gene proteins, which is a novel brain-specific subclass of the lipid phosphate phosphatase superfamily. PLPPR1-5 have prominent roles in synapse formation and axonal pathfinding. We found that PLPPR1 overexpression in the mouse neuroblastoma cell line (Neuro2a) results in increase in cell adhesion and reduced cell migration. During migration, these cells leave behind long fibrous looking extensions of the plasma membrane causing a peculiar phenotype. Cells expressing PLPPR1 showed decreased actin turnover and decreased disassembly of focal adhesions. PLPPR1 also reduced active Rac1, and expressing dominant negative Rac1 produced a similar phenotype to overexpression of PLPPR1. The PLPPR1-induced phenotype of long fibers was reversed by introducing constitutively active Rac1. In summary, we show that PLPPR1 decreases active Rac1 levels that leads to cascade of events which increases cell adhesion.
Collapse
Affiliation(s)
- Sharada Tilve
- Laboratory of Developmental Neurobiology, National Heart Lung and Blood Institute, NIH, Bethesda, MD, 20892, USA
| | - Chinyere Agbaegbu Iweka
- Laboratory of Developmental Neurobiology, National Heart Lung and Blood Institute, NIH, Bethesda, MD, 20892, USA; Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA
| | - Jonathan Bao
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Natalie Hawken
- Laboratory of Developmental Neurobiology, National Heart Lung and Blood Institute, NIH, Bethesda, MD, 20892, USA
| | - Caitlin P Mencio
- Laboratory of Developmental Neurobiology, National Heart Lung and Blood Institute, NIH, Bethesda, MD, 20892, USA
| | - Herbert M Geller
- Laboratory of Developmental Neurobiology, National Heart Lung and Blood Institute, NIH, Bethesda, MD, 20892, USA.
| |
Collapse
|
21
|
Carlier MF. Actin self-assembly: from filament structure and mechanics to motile and morphogenetic processes. Semin Cell Dev Biol 2020; 102:48-50. [PMID: 31926834 DOI: 10.1016/j.semcdb.2020.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
22
|
Abstract
Phagocytosis is a specialized process that enables cellular ingestion and clearance of microbes, dead cells and tissue debris that are too large for other endocytic routes. As such, it is an essential component of tissue homeostasis and the innate immune response, and also provides a link to the adaptive immune response. However, ingestion of large particulate materials represents a monumental task for phagocytic cells. It requires profound reorganization of the cell morphology around the target in a controlled manner, which is limited by biophysical constraints. Experimental and theoretical studies have identified critical aspects associated with the interconnected biophysical properties of the receptors, the membrane, and the actin cytoskeleton that can determine the success of large particle internalization. In this review, we will discuss the major physical constraints involved in the formation of a phagosome. Focusing on two of the most-studied types of phagocytic receptors, the Fcγ receptors and the complement receptor 3 (αMβ2 integrin), we will describe the complex molecular mechanisms employed by phagocytes to overcome these physical constraints.
Collapse
Affiliation(s)
- Valentin Jaumouillé
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Clare M Waterman
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
23
|
De Ieso ML, Pei JV, Nourmohammadi S, Smith E, Chow PH, Kourghi M, Hardingham JE, Yool AJ. Combined pharmacological administration of AQP1 ion channel blocker AqB011 and water channel blocker Bacopaside II amplifies inhibition of colon cancer cell migration. Sci Rep 2019; 9:12635. [PMID: 31477744 PMCID: PMC6718670 DOI: 10.1038/s41598-019-49045-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 08/13/2019] [Indexed: 12/14/2022] Open
Abstract
Aquaporin-1 (AQP1) has been proposed as a dual water and cation channel that when upregulated in cancers enhances cell migration rates; however, the mechanism remains unknown. Previous work identified AqB011 as an inhibitor of the gated human AQP1 cation conductance, and bacopaside II as a blocker of AQP1 water pores. In two colorectal adenocarcinoma cell lines, high levels of AQP1 transcript were confirmed in HT29, and low levels in SW480 cells, by quantitative PCR (polymerase chain reaction). Comparable differences in membrane AQP1 protein levels were demonstrated by immunofluorescence imaging. Migration rates were quantified using circular wound closure assays and live-cell tracking. AqB011 and bacopaside II, applied in combination, produced greater inhibitory effects on cell migration than did either agent alone. The high efficacy of AqB011 alone and in combination with bacopaside II in slowing HT29 cell motility correlated with abundant membrane localization of AQP1 protein. In SW480, neither agent alone was effective in blocking cell motility; however, combined application did cause inhibition of motility, consistent with low levels of membrane AQP1 expression. Bacopaside alone or combined with AqB011 also significantly impaired lamellipodial formation in both cell lines. Knockdown of AQP1 with siRNA (confirmed by quantitative PCR) reduced the effectiveness of the combined inhibitors, confirming AQP1 as a target of action. Invasiveness measured using transwell filters layered with extracellular matrix in both cell lines was inhibited by AqB011, with a greater potency in HT29 than SW480. A side effect of bacopaside II at high doses was a potentiation of invasiveness, that was reversed by AqB011. Results here are the first to demonstrate that combined block of the AQP1 ion channel and water pores is more potent in impairing motility across diverse classes of colon cancer cells than single agents alone.
Collapse
Affiliation(s)
- Michael L De Ieso
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Jinxin V Pei
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Saeed Nourmohammadi
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Eric Smith
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
- Oncology Department, The Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville, SA, 5011, Australia
| | - Pak Hin Chow
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Mohamad Kourghi
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Jennifer E Hardingham
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
- Oncology Department, The Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville, SA, 5011, Australia
| | - Andrea J Yool
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia.
| |
Collapse
|
24
|
Kim M. A numerical mechanical model integrating actin treadmilling and receptor recycling to explain selective disengagement of immune cells. Math Biosci 2019; 316:108244. [PMID: 31449891 DOI: 10.1016/j.mbs.2019.108244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 05/30/2019] [Accepted: 08/23/2019] [Indexed: 11/28/2022]
Abstract
T-killer cells of the immune system eliminate virus-infected cells by releasing toxic granules through a direct cell to cell surface interface (synapse). The release of toxic granules only through the cell surface interface assures the specificity of the immune response. The toxic granule releasing apparatus, however, may not be aligned toward the synapse at the moment of the synapse formation. Therefore, mechanisms for reorienting the killing apparatus inside the T-killer cell to the interface with the target is required. Numerous research works were reported to suggest the mechanisms with direct and indirect evidence, but the most adversary situation, that is when the cell's initial orientation is the complete opposite of the desired direction, either remained answered or brought skepticism toward the suggested mechanisms. To address this issue, a computational mechanical model of T-killer cell synapse formation is constructed to test previously suggested models in a more realistic setting and at the same time to test previously neglected component in the model, namely the actin network formation on the synapse. In this model, by capturing the mechanical interaction between T-killer cell surface receptor dynamics and mechanical properties of synapse formation, it is shown that T-killer cells can selectively engage or disengage from the target cell depending on the killing apparatus alignment with the target. The mechanism works as a safeguard measure ensuring target-cell killing and specificity, so it will be effective when T-killer cells are stranded in situations where the alignment of the killing apparatus is challenging.
Collapse
Affiliation(s)
- Myles Kim
- Department of Mathematics and Statistics, Florida Polytechnic University, Lakeland, FL, United States.
| |
Collapse
|
25
|
Pollard TD. Cell Motility and Cytokinesis: From Mysteries to Molecular Mechanisms in Five Decades. Annu Rev Cell Dev Biol 2019; 35:1-28. [PMID: 31394047 DOI: 10.1146/annurev-cellbio-100818-125427] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This is the story of someone who has been fortunate to work in a field of research where essentially nothing was known at the outset but that blossomed with the discovery of profound insights about two basic biological processes: cell motility and cytokinesis. The field started with no molecules, just a few people, and primitive methods. Over time, technological advances in biophysics, biochemistry, and microscopy allowed the combined efforts of scientists in hundreds of laboratories to explain mysterious processes with molecular mechanisms that can be embodied in mathematical equations and simulated by computers. The success of this field is a tribute to the power of the reductionist strategy for understanding biology.
Collapse
Affiliation(s)
- Thomas D Pollard
- Departments of Molecular, Cellular and Developmental Biology; Molecular Biophysics and Biochemistry; and Cell Biology, Yale University, New Haven, Connecticut 06520-8103, USA;
| |
Collapse
|
26
|
Meenderink LM, Gaeta IM, Postema MM, Cencer CS, Chinowsky CR, Krystofiak ES, Millis BA, Tyska MJ. Actin Dynamics Drive Microvillar Motility and Clustering during Brush Border Assembly. Dev Cell 2019; 50:545-556.e4. [PMID: 31378589 DOI: 10.1016/j.devcel.2019.07.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 05/03/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022]
Abstract
Transporting epithelial cells generate arrays of microvilli, known as a brush border, to enhance functional capacity. To understand brush border formation, we used live cell imaging to visualize apical remodeling early in this process. Strikingly, we found that individual microvilli exhibit persistent active motility, translocating across the cell surface at ∼0.2 μm/min. Perturbation with inhibitors and photokinetic experiments revealed that microvillar motility is driven by actin assembly at the barbed ends of core bundles, which in turn is linked to robust treadmilling of these structures. Actin regulatory factors IRTKS and EPS8 localize to the barbed ends of motile microvilli, where they control the kinetics and nature of movement. As the apical surface of differentiating epithelial cells is crowded with nascent microvilli, persistent motility promotes collisions between protrusions and ultimately clustering and consolidation into higher-order arrays. Thus, microvillar motility represents a previously unrecognized driving force for apical surface remodeling and maturation during epithelial differentiation.
Collapse
Affiliation(s)
- Leslie M Meenderink
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Isabella M Gaeta
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Meagan M Postema
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Caroline S Cencer
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Colbie R Chinowsky
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Evan S Krystofiak
- Cell Imaging Shared Resource, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Bryan A Millis
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Biomedical Engineering, Vanderbilt University School of Engineering, Nashville, TN 37232, USA; Cell Imaging Shared Resource, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Biophotonics Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
27
|
An estimate to the first approximation of microtubule rupture force. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2019; 48:569-577. [PMID: 31134309 DOI: 10.1007/s00249-019-01371-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 02/18/2019] [Accepted: 05/17/2019] [Indexed: 10/26/2022]
Abstract
Microtubule mechanical properties are essential for understanding basic cellular processes, including cell motility and division, but the forces that result in microtubule rupture or breakage have not yet been measured directly. These forces are essential to understand the mechanical properties of the cytoskeleton and responses by cells to both normal conditions and stress caused by injury or disease. Here we estimate the force required to rupture a microtubule by analyzing kinesin-14 Ncd motor-induced microtubule breakage in ensemble motility assays. We model the breakage events as caused by Ncd motors pulling or pushing on single microtubules that are clamped at one end by other motors attached to the glass surface. The number of pulling or pushing Ncd motors is approximated from the length of the microtubule bound to the surface and the forces produced by the pulling or pushing motors are estimated from forces produced by the Ncd motor in laser-trap assays, reported by others. Our analysis provides an estimate, to the first approximation, of ~ 500 pN for the minimal force required to rupture a 13-pf microtubule. The value we report is close to the forces estimated from microtubule stretching/fragmentation experiments and overlaps with the forces applied by AFM in microtubule indentation assays that destabilize microtubules and break microtubule protofilaments. It is also consistent with the forces required to disrupt protein noncovalent bonds in force spectroscopy experiments. These findings are relevant to microtubule deformation and breakage caused by cellular tension in vivo.
Collapse
|
28
|
Effect of mycalolides isolated from a marine sponge Mycale aff. nullarosette on actin in living cells. Sci Rep 2019; 9:7540. [PMID: 31101864 PMCID: PMC6525181 DOI: 10.1038/s41598-019-44036-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 05/08/2019] [Indexed: 11/09/2022] Open
Abstract
Discovery of novel bioactive compounds is important not only for therapeutic purposes but also for understanding the mechanisms of biological processes. To screen bioactive compounds that affect nuclear morphology in marine organism extracts, we employed a microscopy-based assay using DNA staining of human cancer cells. A crude extract from a marine sponge Mycale aff. nullarosette, collected from the east coast of Japan, induced cellular binucleation. Fractionation of the extract led to the isolation of mycalolides A and B, and 38-hydroxymycalolide B as the active components. Mycalolides have been identified as marine toxins that induce depolymerization of the actin filament. Live cell imaging revealed that low concentrations of mycalolide A produce binucleated cells by inhibiting the completion of cytokinesis. At higher concentrations, however, mycalolide A causes immediate disruption of actin filaments and changes in cell morphology, yielding rounded cells. These results suggest that the completion of cytokinesis is a process requiring high actin polymerization activity. Furthermore, luciferase reporter assays with mycalolide A treatments support the view that the level of globular actin can affect transcription of a serum response gene.
Collapse
|
29
|
DuChez BJ, Doyle AD, Dimitriadis EK, Yamada KM. Durotaxis by Human Cancer Cells. Biophys J 2019; 116:670-683. [PMID: 30709621 PMCID: PMC6382956 DOI: 10.1016/j.bpj.2019.01.009] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 12/11/2018] [Accepted: 01/07/2019] [Indexed: 01/05/2023] Open
Abstract
Durotaxis is a type of directed cell migration in which cells respond to a gradient of extracellular stiffness. Using automated tracking of positional data for large sample sizes of single migrating cells, we investigated 1) whether cancer cells can undergo durotaxis; 2) whether cell durotactic efficiency varies depending on the regional compliance of stiffness gradients; 3) whether a specific cell migration parameter such as speed or time of migration correlates with durotaxis; and 4) whether Arp2/3, previously implicated in leading edge dynamics and migration, contributes to cancer cell durotaxis. Although durotaxis has been characterized primarily in nonmalignant mesenchymal cells, little is known about its role in cancer cell migration. Diffusible factors are known to affect cancer cell migration and metastasis. However, because many tumor microenvironments gradually stiffen, we hypothesized that durotaxis might also govern migration of cancer cells. We evaluated the durotactic potential of multiple cancer cell lines by employing substrate stiffness gradients mirroring the physiological stiffness encountered by cells in a variety of tissues. Automated cell tracking permitted rapid acquisition of positional data and robust statistical analyses for migrating cells. These durotaxis assays demonstrated that all cancer cell lines tested (two glioblastoma, metastatic breast cancer, and fibrosarcoma) migrated directionally in response to changes in extracellular stiffness. Unexpectedly, all cancer cell lines tested, as well as noninvasive human fibroblasts, displayed the strongest durotactic migratory response when migrating on the softest regions of stiffness gradients (2-7 kPa), with decreased responsiveness on stiff regions of gradients. Focusing on glioblastoma cells, durotactic forward migration index and displacement rates were relatively stable over time. Correlation analyses showed the expected correlation with displacement along the gradient but much less with persistence and none with cell speed. Finally, we found that inhibition of Arp2/3, an actin-nucleating protein necessary for lamellipodial protrusion, impaired durotactic migration.
Collapse
Affiliation(s)
- Brian J DuChez
- Cell Biology Section, Division of Intramural Research, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Andrew D Doyle
- Cell Biology Section, Division of Intramural Research, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Emilios K Dimitriadis
- Trans-NIH Shared Resource on Biomedical Engineering and Physical Science, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland
| | - Kenneth M Yamada
- Cell Biology Section, Division of Intramural Research, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
30
|
Kollepara KS, Mulye PD, Saez P. Fully coupled numerical model of actin treadmilling in the lamellipodium of the cell. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2018; 34:e3143. [PMID: 30133172 DOI: 10.1002/cnm.3143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 08/06/2018] [Accepted: 08/09/2018] [Indexed: 06/08/2023]
Abstract
Cells rely on an interplay of subcellular elements for motility and migration. Certain regions of motile cells, such as the lamellipodium, are made of a complex mixture of actin monomers and filaments, which polymerize at the front of the cell, close to the cell membrane, and depolymerize at the rear. The dynamic actin turnover induces the so-called intracellular retrograde flow, and it is a fundamental process for cell motility. Apart from some comprehensive mathematical models, the computational modelling of actin treadmilling has been based on simpler biophysical models. Here, we adopt a highly detailed theoretical model of the actin treadmilling process and develop a coupled unsteady finite element formulation. We clearly describe the structure and implementation of the coupled problem within the finite element method. Our numerical results show an excellent correlation with experimental results from literature and with previous models. We include time dependent effects and convective transport terms, which expose puzzling dynamics in the retrograde flow. We propose several biological scenarios to analyze the behavior of the actin treadmilling along space and time. We observed response times of the main density variables in the order of seconds. Compared with previous analytical solutions, which make assumptions related to convective transport, transient dynamics, and actin fluxes, the generic solution can have significant influence on the retrograde flow. All together, our results unveil a promising applicability of classical finite element methods to derive an in silico testing platform for the actin treadmilling processes in motile cells, which could allow for an extension to other biophysical effects.
Collapse
Affiliation(s)
| | - Paris D Mulye
- Ecole Centrale de Nantes, 1 Rue de la Noe, 44300 Nantes, France
| | - Pablo Saez
- Laboratori de Calcul Numeric (LaCaN), Universitat Politécnica de Catalunya-BarcelonaTech (UPC), Barcelona, Spain
| |
Collapse
|
31
|
Higgs HN. A fruitful tree: developing the dendritic nucleation model of actin-based cell motility. Mol Biol Cell 2018. [PMCID: PMC6333179 DOI: 10.1091/mbc.e18-07-0426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
A fundamental question in cell biology concerns how cells move, and this has been the subject of intense research for decades. In the 1990s, a major leap forward was made in our understanding of cell motility, with the proposal of the dendritic nucleation model. This essay describes the events leading to the development of the model, including findings from many laboratories and scientific disciplines. The story is an excellent example of the scientific process in action, with the combination of multiple perspectives leading to robust conclusions.
Collapse
Affiliation(s)
- Henry N. Higgs
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| |
Collapse
|
32
|
Pollard TD. Theory from the Oster Laboratory Leaps Ahead of Experiment in Understanding Actin-Based Cellular Motility. Biophys J 2018; 111:1589-1592. [PMID: 27760345 DOI: 10.1016/j.bpj.2016.08.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 08/30/2016] [Indexed: 10/20/2022] Open
Affiliation(s)
- Thomas D Pollard
- Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, Connecticut; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut; Department of Cell Biology, Yale University, New Haven, Connecticut.
| |
Collapse
|
33
|
Rotation of stress fibers as a single wheel in migrating fish keratocytes. Sci Rep 2018; 8:10615. [PMID: 30018412 PMCID: PMC6050267 DOI: 10.1038/s41598-018-28875-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 06/28/2018] [Indexed: 12/14/2022] Open
Abstract
Crawling migration plays an essential role in a variety of biological phenomena, including development, wound healing, and immune system function. Keratocytes are wound-healing cells in fish skin. Expansion of the leading edge of keratocytes and retraction of the rear are respectively induced by actin polymerization and contraction of stress fibers in the same way as for other cell types. Interestingly, stress fibers in keratocytes align almost perpendicular to the migration-direction. It seems that in order to efficiently retract the rear, it is better that the stress fibers align parallel to it. From the unique alignment of stress fibers in keratocytes, we speculated that the stress fibers may play a role for migration other than the retraction. Here, we reveal that the stress fibers are stereoscopically arranged so as to surround the cytoplasm in the cell body; we directly show, in sequential three-dimensional recordings, their rolling motion during migration. Removal of the stress fibers decreased migration velocity and induced the collapse of the left-right balance of crawling migration. The rotation of these stress fibers plays the role of a “wheel” in crawling migration of keratocytes.
Collapse
|
34
|
De Ieso ML, Yool AJ. Mechanisms of Aquaporin-Facilitated Cancer Invasion and Metastasis. Front Chem 2018; 6:135. [PMID: 29922644 PMCID: PMC5996923 DOI: 10.3389/fchem.2018.00135] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/09/2018] [Indexed: 01/02/2023] Open
Abstract
Cancer is a leading cause of death worldwide, and its incidence is rising with numbers expected to increase 70% in the next two decades. The fact that current mainline treatments for cancer patients are accompanied by debilitating side effects prompts a growing demand for new therapies that not only inhibit growth and proliferation of cancer cells, but also control invasion and metastasis. One class of targets gaining international attention is the aquaporins, a family of membrane-spanning water channels with diverse physiological functions and extensive tissue-specific distributions in humans. Aquaporins−1,−2,−3,−4,−5,−8, and−9 have been linked to roles in cancer invasion, and metastasis, but their mechanisms of action remain to be fully defined. Aquaporins are implicated in the metastatic cascade in processes of angiogenesis, cellular dissociation, migration, and invasion. Cancer invasion and metastasis are proposed to be potentiated by aquaporins in boosting tumor angiogenesis, enhancing cell volume regulation, regulating cell-cell and cell-matrix adhesions, interacting with actin cytoskeleton, regulating proteases and extracellular-matrix degrading molecules, contributing to the regulation of epithelial-mesenchymal transitions, and interacting with signaling pathways enabling motility and invasion. Pharmacological modulators of aquaporin channels are being identified and tested for therapeutic potential, including compounds derived from loop diuretics, metal-containing organic compounds, plant natural products, and other small molecules. Further studies on aquaporin-dependent functions in cancer metastasis are needed to define the differential contributions of different classes of aquaporin channels to regulation of fluid balance, cell volume, small solute transport, signal transduction, their possible relevance as rate limiting steps, and potential values as therapeutic targets for invasion and metastasis.
Collapse
Affiliation(s)
- Michael L De Ieso
- Department of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Andrea J Yool
- Department of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
35
|
Elosegui-Artola A, Trepat X, Roca-Cusachs P. Control of Mechanotransduction by Molecular Clutch Dynamics. Trends Cell Biol 2018; 28:356-367. [PMID: 29496292 DOI: 10.1016/j.tcb.2018.01.008] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/24/2018] [Accepted: 01/30/2018] [Indexed: 02/09/2023]
Abstract
The linkage of cells to their microenvironment is mediated by a series of bonds that dynamically engage and disengage, in what has been conceptualized as the molecular clutch model. Whereas this model has long been employed to describe actin cytoskeleton and cell migration dynamics, it has recently been proposed to also explain mechanotransduction (i.e., the process by which cells convert mechanical signals from their environment into biochemical signals). Here we review the current understanding on how cell dynamics and mechanotransduction are driven by molecular clutch dynamics and its master regulator, the force loading rate. Throughout this Review, we place a specific emphasis on the quantitative prediction of cell response enabled by combined experimental and theoretical approaches.
Collapse
Affiliation(s)
- Alberto Elosegui-Artola
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain.
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; University of Barcelona, 08028 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain; Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, 28029 Madrid, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; University of Barcelona, 08028 Barcelona, Spain.
| |
Collapse
|
36
|
Yun C, Katchko KM, Schallmo MS, Jeong S, Yun J, Chen CH, Weiner JA, Park C, George A, Stupp SI, Hsu WK, Hsu EL. Aryl Hydrocarbon Receptor Antagonists Mitigate the Effects of Dioxin on Critical Cellular Functions in Differentiating Human Osteoblast-Like Cells. Int J Mol Sci 2018; 19:ijms19010225. [PMID: 29324662 PMCID: PMC5796174 DOI: 10.3390/ijms19010225] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/20/2017] [Accepted: 01/10/2018] [Indexed: 01/31/2023] Open
Abstract
The inhibition of bone healing in humans is a well-established effect associated with cigarette smoking, but the underlying mechanisms are still unclear. Recent work using animal cell lines have implicated the aryl hydrocarbon receptor (AhR) as a mediator of the anti-osteogenic effects of cigarette smoke, but the complexity of cigarette smoke mixtures makes understanding the mechanisms of action a major challenge. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD, dioxin) is a high-affinity AhR ligand that is frequently used to investigate biological processes impacted by AhR activation. Since there are dozens of AhR ligands present in cigarette smoke, we utilized dioxin as a prototype ligand to activate the receptor and explore its effects on pro-osteogenic biomarkers and other factors critical to osteogenesis using a human osteoblast-like cell line. We also explored the capacity for AhR antagonists to protect against dioxin action in this context. We found dioxin to inhibit osteogenic differentiation, whereas co-treatment with various AhR antagonists protected against dioxin action. Dioxin also negatively impacted cell adhesion with a corresponding reduction in the expression of integrin and cadherin proteins, which are known to be involved in this process. Similarly, the dioxin-mediated inhibition of cell migration correlated with reduced expression of the chemokine receptor CXCR4 and its ligand, CXCL12, and co-treatment with antagonists restored migratory capacity. Our results suggest that AhR activation may play a role in the bone regenerative response in humans exposed to AhR activators, such as those present in cigarette smoke. Given the similarity of our results using a human cell line to previous work done in murine cells, animal models may yield data relevant to the human setting. In addition, the AhR may represent a potential therapeutic target for orthopedic patients who smoke cigarettes, or those who are exposed to secondhand smoke or other environmental sources of aryl hydrocarbons.
Collapse
Affiliation(s)
- Chawon Yun
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA.
| | - Karina M Katchko
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA.
| | - Michael S Schallmo
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA.
| | - Soyeon Jeong
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA.
| | - Jonghwa Yun
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA.
| | - Charlotte H Chen
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA.
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA.
| | - Joseph A Weiner
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA.
| | - Christian Park
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA.
| | - Andrew George
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA.
| | - Samuel I Stupp
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA.
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA.
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA.
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA.
- Department of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Wellington K Hsu
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA.
| | - Erin L Hsu
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
37
|
Abstract
The actin cytoskeleton-a collection of actin filaments with their accessory and regulatory proteins-is the primary force-generating machinery in the cell. It can produce pushing (protrusive) forces through coordinated polymerization of multiple actin filaments or pulling (contractile) forces through sliding actin filaments along bipolar filaments of myosin II. Both force types are particularly important for whole-cell migration, but they also define and change the cell shape and mechanical properties of the cell surface, drive the intracellular motility and morphogenesis of membrane organelles, and allow cells to form adhesions with each other and with the extracellular matrix.
Collapse
Affiliation(s)
- Tatyana Svitkina
- Department of Biology, University of Pennsylvania, 221 Leidy Labs, Philadelphia, Pennsylvania 19104
| |
Collapse
|
38
|
Raz-Ben Aroush D, Ofer N, Abu-Shah E, Allard J, Krichevsky O, Mogilner A, Keren K. Actin Turnover in Lamellipodial Fragments. Curr Biol 2017; 27:2963-2973.e14. [PMID: 28966086 DOI: 10.1016/j.cub.2017.08.066] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 07/21/2017] [Accepted: 08/29/2017] [Indexed: 10/18/2022]
Abstract
Actin turnover is the central driving force underlying lamellipodial motility. The molecular components involved are largely known, and their properties have been studied extensively in vitro. However, a comprehensive picture of actin turnover in vivo is still missing. We focus on fragments from fish epithelial keratocytes, which are essentially stand-alone motile lamellipodia. The geometric simplicity of the fragments and the absence of additional actin structures allow us to characterize the spatiotemporal lamellipodial actin organization with unprecedented detail. We use fluorescence recovery after photobleaching, fluorescence correlation spectroscopy, and extraction experiments to show that about two-thirds of the lamellipodial actin diffuses in the cytoplasm with nearly uniform density, whereas the rest forms the treadmilling polymer network. Roughly a quarter of the diffusible actin pool is in filamentous form as diffusing oligomers, indicating that severing and debranching are important steps in the disassembly process generating oligomers as intermediates. The remaining diffusible actin concentration is orders of magnitude higher than the in vitro actin monomer concentration required to support the observed polymerization rates, implying that the majority of monomers are transiently kept in a non-polymerizable "reserve" pool. The actin network disassembles and reassembles throughout the lamellipodium within seconds, so the lamellipodial network turnover is local. The diffusible actin transport, on the other hand, is global: actin subunits typically diffuse across the entire lamellipodium before reassembling into the network. This combination of local network turnover and global transport of dissociated subunits through the cytoplasm makes actin transport robust yet rapidly adaptable and amenable to regulation.
Collapse
Affiliation(s)
- Dikla Raz-Ben Aroush
- Department of Physics, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Noa Ofer
- Department of Physics, Technion-Israel Institute of Technology, Haifa 32000, Israel; Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Enas Abu-Shah
- Department of Physics, Technion-Israel Institute of Technology, Haifa 32000, Israel; Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Jun Allard
- Department of Mathematics, Center for Complex Biological Systems and Department of Physics and Astronomy, University of California at Irvine, Irvine, CA 92697, USA
| | - Oleg Krichevsky
- Physics Department and Ilse Kats Center for Nanoscience, Ben-Gurion University, Beer-Sheva 84105, Israel
| | - Alex Mogilner
- Courant Institute of Mathematical Sciences and Department of Biology, New York University, New York, NY 10012, USA.
| | - Kinneret Keren
- Department of Physics, Technion-Israel Institute of Technology, Haifa 32000, Israel; Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 32000, Israel; Network Biology Research Laboratories, Technion-Israel Institute of Technology, Haifa 32000, Israel.
| |
Collapse
|
39
|
Kumar S, Mansson A. Covalent and non-covalent chemical engineering of actin for biotechnological applications. Biotechnol Adv 2017; 35:867-888. [PMID: 28830772 DOI: 10.1016/j.biotechadv.2017.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 08/09/2017] [Accepted: 08/16/2017] [Indexed: 12/26/2022]
Abstract
The cytoskeletal filaments are self-assembled protein polymers with 8-25nm diameters and up to several tens of micrometres length. They have a range of pivotal roles in eukaryotic cells, including transportation of intracellular cargoes (primarily microtubules with dynein and kinesin motors) and cell motility (primarily actin and myosin) where muscle contraction is one example. For two decades, the cytoskeletal filaments and their associated motor systems have been explored for nanotechnological applications including miniaturized sensor systems and lab-on-a-chip devices. Several developments have also revolved around possible exploitation of the filaments alone without their motor partners. Efforts to use the cytoskeletal filaments for applications often require chemical or genetic engineering of the filaments such as specific conjugation with fluorophores, antibodies, oligonucleotides or various macromolecular complexes e.g. nanoparticles. Similar conjugation methods are also instrumental for a range of fundamental biophysical studies. Here we review methods for non-covalent and covalent chemical modifications of actin filaments with focus on critical advantages and challenges of different methods as well as critical steps in the conjugation procedures. We also review potential uses of the engineered actin filaments in nanotechnological applications and in some key fundamental studies of actin and myosin function. Finally, we consider possible future lines of investigation that may be addressed by applying chemical conjugation of actin in new ways.
Collapse
Affiliation(s)
- Saroj Kumar
- Department of Biotechnology, Delhi Technological University, Delhi 110042, India; Department of Chemistry and Biomedical Sciences, Faculty of Health and Life Sciences, Linnaeus University, SE-391 82 Kalmar, Sweden.
| | - Alf Mansson
- Department of Chemistry and Biomedical Sciences, Faculty of Health and Life Sciences, Linnaeus University, SE-391 82 Kalmar, Sweden.
| |
Collapse
|
40
|
Lemon DJ, Yang X, Srivastava P, Luk YY, Garza AG. Polymertropism of rod-shaped bacteria: movement along aligned polysaccharide fibers. Sci Rep 2017; 7:7643. [PMID: 28801641 PMCID: PMC5554183 DOI: 10.1038/s41598-017-07486-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 06/16/2017] [Indexed: 01/11/2023] Open
Abstract
In nature, bacteria often live in surface-associated communities known as biofilms. Biofilm-forming bacteria typically deposit a layer of polysaccharide on the surfaces they inhabit; hence, polysaccharide is their immediate environment on many surfaces. In this study, we examined how the physical characteristics of polysaccharide substrates influence the behavior of the biofilm-forming bacterium Myxococcus xanthus. M. xanthus responds to the compression-induced deformation of polysaccharide substrates by preferentially spreading across the surface perpendicular to the axis of compression. Our results suggest that M. xanthus is not responding to the water that accumulates on the surface of the polysaccharide substrate after compression or to compression-induced changes in surface topography such as the formation of troughs. These directed surface movements do, however, consistently match the orientation of the long axes of aligned and tightly packed polysaccharide fibers in compressed substrates, as indicated by behavioral, birefringence and small angle X-ray scattering analyses. Therefore, we suggest that the directed movements are a response to the physical arrangement of the polymers in the substrate and refer to the directed movements as polymertropism. This behavior might be a common property of bacteria, as many biofilm-forming bacteria that are rod-shaped and motile on soft surfaces exhibit polymertropism.
Collapse
Affiliation(s)
- David J Lemon
- Department of Biology, Syracuse University, Syracuse, NY, 13244, United States
| | - Xingbo Yang
- Department of Physics, Syracuse University, Syracuse, NY, 13244, United States.,Department of Physics and Astronomy, Northwestern University, Evanston, IL, 60208, United States
| | - Pragya Srivastava
- Department of Physics, Syracuse University, Syracuse, NY, 13244, United States.,The Francis Crick Institute, London, NW1 1BF, United Kingdom
| | - Yan-Yeung Luk
- Department of Chemistry, Syracuse University, Syracuse, NY, 13244, United States.
| | - Anthony G Garza
- Department of Biology, Syracuse University, Syracuse, NY, 13244, United States.
| |
Collapse
|
41
|
Yamashiro S, Watanabe N. Overview of Single-Molecule Speckle (SiMS) Microscopy and Its Electroporation-Based Version with Efficient Labeling and Improved Spatiotemporal Resolution. SENSORS 2017; 17:s17071585. [PMID: 28684722 PMCID: PMC5539652 DOI: 10.3390/s17071585] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 12/19/2022]
Abstract
Live-cell single-molecule imaging was introduced more than a decade ago, and has provided critical information on remodeling of the actin cytoskeleton, the motion of plasma membrane proteins, and dynamics of molecular motor proteins. Actin remodeling has been the best target for this approach because actin and its associated proteins stop diffusing when assembled, allowing visualization of single-molecules of fluorescently-labeled proteins in a state specific manner. The approach based on this simple principle is called Single-Molecule Speckle (SiMS) microscopy. For instance, spatiotemporal regulation of actin polymerization and lifetime distribution of actin filaments can be monitored directly by tracking actin SiMS. In combination with fluorescently labeled probes of various actin regulators, SiMS microscopy has contributed to clarifying the processes underlying recycling, motion and remodeling of the live-cell actin network. Recently, we introduced an electroporation-based method called eSiMS microscopy, with high efficiency, easiness and improved spatiotemporal precision. In this review, we describe the application of live-cell single-molecule imaging to cellular actin dynamics and discuss the advantages of eSiMS microscopy over previous SiMS microscopy.
Collapse
Affiliation(s)
- Sawako Yamashiro
- Laboratory of Single-Molecule Cell Biology, Kyoto University Graduate School of Biostudies, Kyoto 606-8501, Japan.
| | - Naoki Watanabe
- Laboratory of Single-Molecule Cell Biology, Kyoto University Graduate School of Biostudies, Kyoto 606-8501, Japan.
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan.
| |
Collapse
|
42
|
Bachir AI, Horwitz AR, Nelson WJ, Bianchini JM. Actin-Based Adhesion Modules Mediate Cell Interactions with the Extracellular Matrix and Neighboring Cells. Cold Spring Harb Perspect Biol 2017; 9:9/7/a023234. [PMID: 28679638 DOI: 10.1101/cshperspect.a023234] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cell adhesions link cells to the extracellular matrix (ECM) and to each other and depend on interactions with the actin cytoskeleton. Both cell-ECM and cell-cell adhesion sites contain discrete, yet overlapping, functional modules. These modules establish physical associations with the actin cytoskeleton, locally modulate actin organization and dynamics, and trigger intracellular signaling pathways. Interplay between these modules generates distinct actin architectures that underlie different stages, types, and functions of cell-ECM and cell-cell adhesions. Actomyosin contractility is required to generate mature, stable adhesions, as well as to sense and translate the mechanical properties of the cellular environment into changes in cell organization and behavior. Here, we review the organization and function of different adhesion modules and how they interact with the actin cytoskeleton. We highlight the molecular mechanisms of mechanotransduction in adhesions and how adhesion molecules mediate cross talk between cell-ECM and cell-cell adhesion sites.
Collapse
Affiliation(s)
- Alexia I Bachir
- Protein and Cell Analysis, Biosciences Division, Thermo Fisher Scientific, Eugene, Oregon 97402
| | - Alan Rick Horwitz
- Protein and Cell Analysis, Biosciences Division, Thermo Fisher Scientific, Eugene, Oregon 97402
| | - W James Nelson
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22903
| | - Julie M Bianchini
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22903
| |
Collapse
|
43
|
Schiffhauer ES, Robinson DN. Mechanochemical Signaling Directs Cell-Shape Change. Biophys J 2017; 112:207-214. [PMID: 28122209 DOI: 10.1016/j.bpj.2016.12.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 11/07/2016] [Accepted: 12/01/2016] [Indexed: 12/19/2022] Open
Abstract
For specialized cell function, as well as active cell behaviors such as division, migration, and tissue development, cells must undergo dynamic changes in shape. To complete these processes, cells integrate chemical and mechanical signals to direct force production. This mechanochemical integration allows for the rapid production and adaptation of leading-edge machinery in migrating cells, the invasion of one cell into another during cell-cell fusion, and the force-feedback loops that ensure robust cytokinesis. A quantitative understanding of cell mechanics coupled with protein dynamics has allowed us to account for furrow ingression during cytokinesis, a model cell-shape-change process. At the core of cell-shape changes is the ability of the cell's machinery to sense mechanical forces and tune the force-generating machinery as needed. Force-sensitive cytoskeletal proteins, including myosin II motors and actin cross-linkers such as α-actinin and filamin, accumulate in response to internally generated and externally imposed mechanical stresses, endowing the cell with the ability to discern and respond to mechanical cues. The physical theory behind how these proteins display mechanosensitive accumulation has allowed us to predict paralog-specific behaviors of different cross-linking proteins and identify a zone of optimal actin-binding affinity that allows for mechanical stress-induced protein accumulation. These molecular mechanisms coupled with the mechanical feedback systems ensure robust shape changes, but if they go awry, they are poised to promote disease states such as cancer cell metastasis and loss of tissue integrity.
Collapse
Affiliation(s)
- Eric S Schiffhauer
- Department of Cell Biology, Johns Hopkins University, Baltimore, Maryland
| | - Douglas N Robinson
- Department of Cell Biology, Johns Hopkins University, Baltimore, Maryland; Department of Pharmacology and Molecular Science, Johns Hopkins University, Baltimore, Maryland; Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
44
|
Kage F, Winterhoff M, Dimchev V, Mueller J, Thalheim T, Freise A, Brühmann S, Kollasser J, Block J, Dimchev G, Geyer M, Schnittler HJ, Brakebusch C, Stradal TEB, Carlier MF, Sixt M, Käs J, Faix J, Rottner K. FMNL formins boost lamellipodial force generation. Nat Commun 2017; 8:14832. [PMID: 28327544 PMCID: PMC5364437 DOI: 10.1038/ncomms14832] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 02/03/2017] [Indexed: 01/16/2023] Open
Abstract
Migration frequently involves Rac-mediated protrusion of lamellipodia, formed by Arp2/3 complex-dependent branching thought to be crucial for force generation and stability of these networks. The formins FMNL2 and FMNL3 are Cdc42 effectors targeting to the lamellipodium tip and shown here to nucleate and elongate actin filaments with complementary activities in vitro. In migrating B16-F1 melanoma cells, both formins contribute to the velocity of lamellipodium protrusion. Loss of FMNL2/3 function in melanoma cells and fibroblasts reduces lamellipodial width, actin filament density and -bundling, without changing patterns of Arp2/3 complex incorporation. Strikingly, in melanoma cells, FMNL2/3 gene inactivation almost completely abolishes protrusion forces exerted by lamellipodia and modifies their ultrastructural organization. Consistently, CRISPR/Cas-mediated depletion of FMNL2/3 in fibroblasts reduces both migration and capability of cells to move against viscous media. Together, we conclude that force generation in lamellipodia strongly depends on FMNL formin activity, operating in addition to Arp2/3 complex-dependent filament branching.
Collapse
Affiliation(s)
- Frieda Kage
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany.,Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Moritz Winterhoff
- Institute for Biophysical Chemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Vanessa Dimchev
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany.,Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Jan Mueller
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Tobias Thalheim
- Soft Matter Physics Group, Institut für experimentelle Physik I, Leipzig University, Linnéstraße 5, 04103 Leipzig, Germany
| | - Anika Freise
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany.,Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Stefan Brühmann
- Institute for Biophysical Chemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Jana Kollasser
- Biomedical Institute, BRIC, University of Copenhagen, DK-2200 Copenhagen, Denmark.,Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Jennifer Block
- Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Georgi Dimchev
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany.,Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Matthias Geyer
- Institute of Innate Immunity, Department of Structural Immunology, University of Bonn, Sigmund-Freud-Strasse 25, 53127 Bonn, Germany
| | - Hans-Joachim Schnittler
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149 Münster, Germany
| | - Cord Brakebusch
- Biomedical Institute, BRIC, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Theresia E B Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Marie-France Carlier
- Cytoskeleton Dynamics and Motility Group, Laboratoire d'Enzymologie et Biochimie Structurales, Centre de Recherche de Gif, CNRS, Gif-sur-Yvette 91198, France
| | - Michael Sixt
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Josef Käs
- Soft Matter Physics Group, Institut für experimentelle Physik I, Leipzig University, Linnéstraße 5, 04103 Leipzig, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany.,Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| |
Collapse
|
45
|
Dimchev G, Steffen A, Kage F, Dimchev V, Pernier J, Carlier MF, Rottner K. Efficiency of lamellipodia protrusion is determined by the extent of cytosolic actin assembly. Mol Biol Cell 2017; 28:1311-1325. [PMID: 28331069 PMCID: PMC5426846 DOI: 10.1091/mbc.e16-05-0334] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 02/10/2017] [Accepted: 03/17/2017] [Indexed: 11/26/2022] Open
Abstract
Lamellipodia protrusion requires actin network formation driven by the Arp2/3 complex and its upstream regulators WAVE complex and Rac. Actin assembly factors of the formin and Ena/VASP families can influence protrusion, in particular by maintaining a balance between lamellipodial and cytosolic actin filament assembly. Cell migration and cell–cell communication involve the protrusion of actin-rich cell surface projections such as lamellipodia and filopodia. Lamellipodia are networks of actin filaments generated and turned over by filament branching through the Arp2/3 complex. Inhibition of branching is commonly agreed to eliminate formation and maintenance of lamellipodial actin networks, but the regulation of nucleation or elongation of Arp2/3-independent filament populations within the network by, for example, formins or Ena/VASP family members and its influence on the effectiveness of protrusion have been unclear. Here we analyzed the effects of a set of distinct formin fragments and VASP on site-specific, lamellipodial versus cytosolic actin assembly and resulting consequences on protrusion. Surprisingly, expression of formin variants but not VASP reduced lamellipodial protrusion in B16-F1 cells, albeit to variable extents. The rates of actin network polymerization followed a similar trend. Unexpectedly, the degree of inhibition of both parameters depended on the extent of cytosolic but not lamellipodial actin assembly. Indeed, excess cytosolic actin assembly prevented actin monomer from rapid translocation to and efficient incorporation into lamellipodia. Thus, as opposed to sole regulation by actin polymerases operating at their tips, the protrusion efficiency of lamellipodia is determined by a finely tuned balance between lamellipodial and cytosolic actin assembly.
Collapse
Affiliation(s)
- Georgi Dimchev
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Anika Steffen
- Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Frieda Kage
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Vanessa Dimchev
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Julien Pernier
- Cytoskeleton Dynamics and Motility, Centre National de la Recherche Scientifique, 91198 Gif-sur-Yvette, France
| | - Marie-France Carlier
- Cytoskeleton Dynamics and Motility, Centre National de la Recherche Scientifique, 91198 Gif-sur-Yvette, France
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany .,Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| |
Collapse
|
46
|
Hohmann T, Grabiec U, Ghadban C, Feese K, Dehghani F. The influence of biomechanical properties and cannabinoids on tumor invasion. Cell Adh Migr 2017; 11:54-67. [PMID: 27149140 PMCID: PMC5308229 DOI: 10.1080/19336918.2016.1183867] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 03/10/2016] [Accepted: 04/25/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Cannabinoids are known to have an anti-tumorous effect, but the underlying mechanisms are only sparsely understood. Mechanical characteristics of tumor cells represent a promising marker to distinguish between tumor cells and the healthy tissue. We tested the hypothesis whether cannabinoids influence the tumor cell specific mechanical and migratory properties and if these factors are a prognostic marker for the invasiveness of tumor cells. METHODS 3 different glioblastoma cell lines were treated with cannabinoids and changes of mechanical and migratory properties of single cells were measured using atomic force microscopy and time lapse imaging. The invasiveness of cell lines was determined using a co-culture model with organotypic hippocampal slice cultures. RESULTS We found that cannabinoids are capable of influencing migratory and mechanical properties in a cell line specific manner. A network analysis revealed a correlation between a "generalized stiffness" and the invasiveness for all tumor cell lines after 3 and 4 d of invasion time: r3d = -0.88 [-0.52;-0.97]; r4d = -0.90 [-0.59;-0.98]. CONCLUSIONS Here we could show that a "generalized stiffness" is a profound marker for the invasiveness of a tumor cell population in our model and thus might be of high clinical relevance for drug testing. Additionally cannabinoids were shown to be of potential use for therapeutic approaches of glioblastoma.
Collapse
Affiliation(s)
- Tim Hohmann
- Department of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Urszula Grabiec
- Department of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Chalid Ghadban
- Department of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Kerstin Feese
- Department of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Faramarz Dehghani
- Department of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
47
|
Singh PP, Hawthorne JL, Davis CA, Quintero OA. Permeabilization activated reduction in fluorescence: A novel method to measure kinetics of protein interactions with intracellular structures. Cytoskeleton (Hoboken) 2016; 73:271-85. [PMID: 27126922 DOI: 10.1002/cm.21306] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 12/31/2022]
Abstract
Understanding kinetic information is fundamental in understanding biological function. Advanced imaging technologies have fostered the development of kinetic analyses in cells. We have developed Permeabilization Activated Reduction in Fluorescence (PARF) analysis for determination of apparent t1/2 and immobile fraction, describing the dissociation of a protein of interest from intracellular structures. To create conditions where dissociation events are observable, cells expressing a fluorescently-tagged protein are permeabilized with digitonin, diluting the unbound protein into the extracellular media. As the media volume is much larger than the cytosolic volume, the concentration of the unbound pool decreases drastically, shifting the system out of equilibrium, favoring dissociation events. Loss of bound protein is observed as loss of fluorescence from intracellular structures and can be fit to an exponential decay. We compared PARF dissociation kinetics with previously published equilibrium kinetics as determined by FRAP. PARF dissociation rates agreed with the equilibrium-based FRAP analysis predictions of the magnitude of those rates. When used to investigate binding kinetics of a panel of cytoskeletal proteins, PARF analysis revealed that filament stabilization resulted in slower fluorescence loss. Additionally, commonly used "general" F-actin labels display differences in kinetic properties, suggesting that not all fluorescently-tagged actin labels interact with the actin network in the same way. We also observed differential dissociation kinetics for GFP-VASP depending on which cellular structure was being labeled. These results demonstrate that PARF analysis of non-equilibrium systems reveals kinetic information without the infrastructure investment required for other quantitative approaches such as FRAP, photoactivation, or in vitro reconstitution assays. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Pali P Singh
- Department of Biology, University of Richmond, Richmond, Virginia
| | | | - Christie A Davis
- Department of Biology, University of Richmond, Richmond, Virginia
| | - Omar A Quintero
- Department of Biology, University of Richmond, Richmond, Virginia
| |
Collapse
|
48
|
Identification of a shootin1 isoform expressed in peripheral tissues. Cell Tissue Res 2016; 366:75-87. [PMID: 27177867 DOI: 10.1007/s00441-016-2415-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/20/2016] [Indexed: 10/24/2022]
Abstract
Shootin1 is a brain-specific cytoplasmic protein involved in neuronal polarity formation and axon outgrowth. It accumulates at the leading edge of axonal growth cones, where it mediates the mechanical coupling between F-actin retrograde flow and cell adhesions as a clutch molecule, thereby producing force for axon outgrowth. In this study, we report a novel splicing isoform of shootin1 which is expressed not only in the brain but also in peripheral tissues. We have renamed the brain-specific shootin1 as shootin1a and termed the novel isoform as shootin1b. Immunoblot and immunohistochemical analyses with a shootin1b-specific antibody revealed that shootin1b is distributed in various mouse tissues including the lung, liver, stomach, intestines, spleen, pancreas, kidney and skin. Interestingly, shootin1b immunoreactivity was widely detected in epithelial cells that constitute simple and stratified epithelia; in some cells, it colocalized with E-cadherin and cortactin at cell-cell contact sites. Shootin1b also localized in dendritic cells in the spleen. These results suggest that shootin1b may function in various peripheral tissues including epithelial cells.
Collapse
|
49
|
Siton-Mendelson O, Bernheim-Groswasser A. Toward the reconstitution of synthetic cell motility. Cell Adh Migr 2016; 10:461-474. [PMID: 27019160 DOI: 10.1080/19336918.2016.1170260] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cellular motility is a fundamental process essential for embryonic development, wound healing, immune responses, and tissues development. Cells are mostly moving by crawling on external, or inside, substrates which can differ in their surface composition, geometry, and dimensionality. Cells can adopt different migration phenotypes, e.g., bleb-based and protrusion-based, depending on myosin contractility, surface adhesion, and cell confinement. In the few past decades, research on cell motility has focused on uncovering the major molecular players and their order of events. Despite major progresses, our ability to infer on the collective behavior from the molecular properties remains a major challenge, especially because cell migration integrates numerous chemical and mechanical processes that are coupled via feedbacks that span over large range of time and length scales. For this reason, reconstituted model systems were developed. These systems allow for full control of the molecular constituents and various system parameters, thereby providing insight into their individual roles and functions. In this review we describe the various reconstituted model systems that were developed in the past decades. Because of the multiple steps involved in cell motility and the complexity of the overall process, most of the model systems focus on very specific aspects of the individual steps of cell motility. Here we describe the main advancement in cell motility reconstitution and discuss the main challenges toward the realization of a synthetic motile cell.
Collapse
Affiliation(s)
- Orit Siton-Mendelson
- a Department of Chemical Engineering and the Ilse Kats Institute for Nanoscale Science and Technology , Ben-Gurion University of the Negev , Beer-Sheva , Israel
| | - Anne Bernheim-Groswasser
- a Department of Chemical Engineering and the Ilse Kats Institute for Nanoscale Science and Technology , Ben-Gurion University of the Negev , Beer-Sheva , Israel
| |
Collapse
|
50
|
Huang SH, Law CH, Kuo PH, Hu RY, Yang CC, Chung TW, Li JM, Lin LH, Liu YC, Liao EC, Tsai YT, Wei YS, Lin CC, Chang CW, Chou HC, Wang WC, Chang MDT, Wang LH, Kung HJ, Chan HL, Lyu PC. MMP-13 is involved in oral cancer cell metastasis. Oncotarget 2016; 7:17144-61. [PMID: 26958809 PMCID: PMC4941377 DOI: 10.18632/oncotarget.7942] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 12/31/1969] [Indexed: 12/20/2022] Open
Abstract
The oral cancer cell line OC3-I5 with a highly invasive ability was selected and derived from an established OSCC line OC3. In this study, we demonstrated that matrix metalloproteinases protein MMP-13 was up-regulated in OC3-I5 than in OC3 cells. We also observed that expression of epithelial-mesenchymal transition (EMT) markers including Twist, p-Src, Snail1, SIP1, JAM-A, and vinculin were increased in OC3-I5 compared to OC3 cells, whereas E-cadherin expression was decreased in the OC3-I5 cells. Using siMMP-13 knockdown techniques, we showed that siMMP-13 not only reduced the invasion and migration, but also the adhesion abilities of oral cancer cells. In support of the role of MMP-13 in metastasis, we used MMP-13 expressing plasmid-transfected 293T cells to enhance MMP-13 expression in the OC3 cells, transplanting the MMP-13 over expressing OC3 cells into nude mice led to enhanced lung metastasis. In summary, our findings show that MMP-13 promotes invasion and metastasis in oral cancer cells, suggesting altered expression of MMP-13 may be utilized to impede the process of metastasis.
Collapse
Affiliation(s)
- Shun-Hong Huang
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Hsuan Law
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ping-Hsueh Kuo
- Department of Medical Sciences and Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ren-Yu Hu
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Chieh Yang
- Department of Radiation Oncology, Chi-Mei Medical Center, Tainan, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Ting-Wen Chung
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ji-Min Li
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Li-Hsun Lin
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Chung Liu
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan.,Institute of Population Sciences, National Health Research Institutes, Miaoli County, Taiwan
| | - En-Chi Liao
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Ting Tsai
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Shan Wei
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chi-Chen Lin
- Institute of Biomedical Science, National Chung Hsing University, Taichung, Taiwan.,Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.,Department of Medical Research and Education, Taichung Veterans General Hospital, Taichung, Taiwan.,Division of Chest Medicine. Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Chien-Wen Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Taiwan
| | - Hsiu-Chuan Chou
- Department of Applied Science, National Hsinchu University of Education, Hsinchu, Taiwan
| | - Wen-Ching Wang
- Department of Medical Sciences and Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Margaret Dah-Tsyr Chang
- Department of Medical Sciences and Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Lu-Hai Wang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Hsing-Jien Kung
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Hong-Lin Chan
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan.,Department of Medical Sciences and Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ping-Chiang Lyu
- Department of Medical Sciences and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan.,Department of Medical Sciences and Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|